Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34953135

RESUMO

Hypothalamic kisspeptin (Kiss1) neurons provide indispensable excitatory transmission to gonadotropin-releasing hormone (GnRH) neurons for the coordinated release of gonadotropins, estrous cyclicity, and ovulation. But maintaining reproductive functions is metabolically demanding so there must be a coordination with multiple homeostatic functions, and it is apparent that Kiss1 neurons play that role. There are 2 distinct populations of hypothalamic Kiss1 neurons, namely arcuate nucleus (Kiss1ARH) neurons and anteroventral periventricular and periventricular nucleus (Kiss1AVPV/PeN) neurons in rodents, both of which excite GnRH neurons via kisspeptin release but are differentially regulated by ovarian steroids. Estradiol (E2) increases the expression of kisspeptin in Kiss1AVPV/PeN neurons but decreases its expression in Kiss1ARH neurons. Also, Kiss1ARH neurons coexpress glutamate and Kiss1AVPV/PeN neurons coexpress gamma aminobutyric acid (GABA), both of which are upregulated by E2 in females. Also, Kiss1ARH neurons express critical metabolic hormone receptors, and these neurons are excited by insulin and leptin during the fed state. Moreover, Kiss1ARH neurons project to and excite the anorexigenic proopiomelanocortin neurons but inhibit the orexigenic neuropeptide Y/Agouti-related peptide neurons, highlighting their role in regulating feeding behavior. Kiss1ARH and Kiss1AVPV/PeN neurons also project to the preautonomic paraventricular nucleus (satiety) neurons and the dorsomedial nucleus (energy expenditure) neurons to differentially regulate their function via glutamate and GABA release, respectively. Therefore, this review will address not only how Kiss1 neurons govern GnRH release, but how they control other homeostatic functions through their peptidergic, glutamatergic and GABAergic synaptic connections, providing further evidence that Kiss1 neurons are the key neurons coordinating energy states with reproduction.


Assuntos
Homeostase/fisiologia , Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Animais , Regulação da Temperatura Corporal , Química Encefálica , Metabolismo Energético/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Kisspeptinas/análise , Kisspeptinas/genética , Hormônio Luteinizante/metabolismo , RNA Mensageiro/análise , Reprodução/fisiologia
2.
Reprod Biol ; 20(4): 474-483, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32807716

RESUMO

Infertility is a growing worldwide public health problem, and stress is a main factor exerting detrimental effects on female reproduction. However, knowledge regarding the neuroendocrine changes caused by chronic stress in females is limited. Therefore, this study assessed the effects of stress on hormones that control female reproduction during the proestrus and diestrus stages of the estrous cycle, as well as its effects on fertility. Adult females were assigned to either a control or a stress group. Stress consisted of exposure, for 15 min, to cold-water immersion daily for 30 days. Estrous cyclicity, female sexual behavior, as well as hypothalamic kisspeptin, gonadotropin releasing hormone (GnRH) content, serum luteinizing hormone (LH), estradiol (E2), progesterone (P4), corticosterone (CORT) and fertility were assessed after chronic stress. The results show that chronically stressed females exhibited disrupted estrous cyclicity, decreased receptivity, low pregnancy rates and lower numbers of fetuses. The content of Kisspeptin and GnRH in the Anteroventral Periventricular/medial Preoptic Area decreased during proestrus, while Kisspeptin increased in the Arcuate nucleus in proestrus and diestrus. Serum LH decreased only during proestrus, whereas E2 and P4 concentrations decreased during proestrus and diestrus, with a concomitant increase in CORT levels in both stages. As a whole, these results indicate that chronic stress decreases Kisspeptin content in AVPV nucleus and GnRH in POA in females, and might induce disruption of the LH surge, consequently disrupting estrous cyclicity and fertility, leading to lower rates of pregnancy and number of fetuses.


Assuntos
Infertilidade Feminina/etiologia , Sistemas Neurossecretores/fisiopatologia , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia , Animais , Corticosterona/sangue , Estradiol/sangue , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/análise , Hipotálamo/química , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/psicologia , Kisspeptinas/análise , Hormônio Luteinizante/sangue , Progesterona/sangue , Ratos , Ratos Wistar , Comportamento Sexual Animal
3.
J Chin Med Assoc ; 83(11): 1020-1028, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32732529

RESUMO

BACKGROUND: This study evaluated whether obese male mice exposed to di-(2-ethylhexyl) phthalate (DEHP) showed synergistic effects on testosterone levels and the potential underlying mechanism. METHODS: Forty-eight male mice were assigned to six groups for 12-week treatments as follows: normal, DEHP100, diet-induced obesity (DIO), DIO + DEHP30, DIO + DEHP100, and DIO + DEHP300. Serum hormone levels, including testosterone (T), luteinizing hormone (LH), and leptin, were detected by ELISA. The levels of Ob-R, kisspeptin, and GPR54 protein expression in hypothalamus and testicular tissues were measured by western blot. RESULTS: There were significantly lower levels of serum T and LH, higher levels of serum leptin and Ob-R, and kisspeptin and GPR54 protein expression were reduced in hypothalamus and testicular tissues in the DIO and DEHP groups compared with controls. Moreover, serum T and leptin levels were more severe in the combined DIO and DEHP exposure group than in the single exposure groups. Serum LH levels and GPR54 expression in the testis were significantly decreased in DIO + DEHP300 mice compared with DIO mice (p < 0.05). CONCLUSION: Obesity- and DEHP-only exposure had adverse effects on testosterone levels in mice, which may be due to high leptin levels and decreased Ob-R, kisspeptin, and GPR54 expression. Obesity combined with DEHP exposure had an additive adverse effect on testosterone levels in mice. One of the potential mechanisms is higher leptin levels and decreased GPR54 expression in the testes.


Assuntos
Dietilexilftalato/toxicidade , Hipotálamo/metabolismo , Kisspeptinas/análise , Obesidade/metabolismo , Receptores de Kisspeptina-1/análise , Testículo/metabolismo , Testosterona/sangue , Animais , Leptina/sangue , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Biol Reprod ; 103(1): 49-59, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32307518

RESUMO

Kisspeptin has been implicated in the ovulatory process of several species of spontaneous ovulators but in only one induced ovulator. In contrast, NGF in semen is the principal trigger of ovulation in other species of induced ovulators-camelids. We tested the hypotheses that kisspeptin induces luteinizing hormone (LH) secretion in llamas through a hypothalamic mechanism, and kisspeptin neurons are the target of NGF in its ovulation-inducing pathway. In Experiment 1, llamas were given either NGF, kisspeptin, or saline intravenously, and LH secretion and ovulation were compared among groups. All llamas treated with NGF (5/5) or kisspeptin (5/5) had an elevation of LH blood concentrations after treatment and ovulated, whereas none of the saline group did (0/5). In Experiment 2, llamas were either pretreated with a gonadotropin-releasing hormone (GnRH) receptor antagonist or saline and treated 2 h later with kisspeptin. Llamas pretreated with saline had elevated plasma LH concentrations and ovulated (6/6) whereas llamas pretreated with cetrorelix did not (0/6). In Experiment 3, we evaluated the hypothalamic kisspeptin-GnRH neuronal network by immunohistochemistry. Kisspeptin neurons were detected in the arcuate nucleus, the preoptic area, and the anterior hypothalamus, establishing synaptic contacts with GnRH neurons. We found no colocalization between kisspeptin and NGF receptors by double immunofluorescence. Functional and morphological findings support the concept that kisspeptin is a mediator of the LH secretory pathway in llamas; however, the role of kisspeptins in the NGF ovulation-inducing pathway in camelids remains unclear since NGF receptors were not detected in kisspeptin neurons in the hypothalamus.


Assuntos
Camelídeos Americanos/fisiologia , Kisspeptinas/farmacologia , Hormônio Luteinizante/metabolismo , Indução da Ovulação/veterinária , Ovulação/efeitos dos fármacos , Ovulação/fisiologia , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/química , Kisspeptinas/análise , Kisspeptinas/fisiologia , Masculino , Fator de Crescimento Neural/isolamento & purificação , Fator de Crescimento Neural/farmacologia , Neurônios/química , Receptores de Fator de Crescimento Neural/análise , Sêmen/química
5.
J Comp Neurol ; 528(1): 32-47, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31251823

RESUMO

The dromedary camel (Camelus dromedarius) is a desert mammal whose cycles in reproductive activity ensure that the offspring's birth and weaning coincide with periods of abundant food resources and favorable climate conditions. In this study, we assessed whether kisspeptin (Kp) and arginine-phenylalanine (RF)-amide related peptide-3 (RFRP-3), two hypothalamic peptides known to regulate the mammalian hypothalamo-pituitary gonadal axis, may be involved in the seasonal control of camel's reproduction. Using specific antibodies and riboprobes, we found that Kp neurons are present in the preoptic area (POA), suprachiasmatic (SCN), and arcuate (ARC) nuclei, and that RFRP-3 neurons are present in the paraventricular (PVN), dorsomedial (DMH), and ventromedial (VMH) hypothalamic nuclei. Kp fibers are found in various hypothalamic areas, notably the POA, SCN, PVN, DMH, VMH, supraoptic nucleus, and the ventral and dorsal premammillary nucleus. RFRP-3 fibers are found in the POA, SCN, PVN, DMH, VMH, and ARC. POA and ARC Kp neurons and DMH RFRP-3 neurons display sexual dimorphism with more neurons in female than in male. Both neuronal populations display opposed seasonal variations with more Kp neurons and less RFRP-3 neurons during the breeding (December-January) than the nonbreeding (July-August) season. This study is the first describing Kp and RFRP-3 in the camel's brain with, during the winter period lower RFRP-3 expression and higher Kp expression possibly responsible for the HPG axis activation. Altogether, our data indicate the involvement of both Kp and RFRP-3 in the seasonal control of the dromedary camel's breeding activity.


Assuntos
Cruzamento , Camelus/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuropeptídeos/metabolismo , Estações do Ano , Sequência de Aminoácidos , Animais , Camelus/genética , Feminino , Hipotálamo/química , Kisspeptinas/análise , Kisspeptinas/genética , Masculino , Neuropeptídeos/análise , Neuropeptídeos/genética , Coelhos , Caracteres Sexuais
6.
Elife ; 82019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30946012

RESUMO

The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Estradiol induces negative feedback on pulsatile GnRH/luteinizing hormone (LH) release and positive feedback generating preovulatory GnRH/LH surges. Negative and positive feedbacks are postulated to be mediated by kisspeptin neurons in arcuate and anteroventral periventricular (AVPV) nuclei, respectively. Kisspeptin-specific ERα knockout mice exhibit disrupted LH pulses and surges. This knockout approach is neither location-specific nor temporally controlled. We utilized CRISPR-Cas9 to disrupt ERα in adulthood. Mice with ERα disruption in AVPV kisspeptin neurons have typical reproductive cycles but blunted LH surges, associated with decreased excitability of these neurons. Mice with ERα knocked down in arcuate kisspeptin neurons showed disrupted cyclicity, associated with increased glutamatergic transmission to these neurons. These observations suggest that activational effects of estradiol regulate surge generation and maintain cyclicity through AVPV and arcuate kisspeptin neurons, respectively, independent from its role in the development of hypothalamic kisspeptin neurons or puberty onset.


Assuntos
Hipotálamo/fisiologia , Neurônios/fisiologia , Reprodução , Comportamento Sexual Animal , Animais , Estradiol/metabolismo , Receptor alfa de Estrogênio/deficiência , Feminino , Técnicas de Inativação de Genes , Kisspeptinas/análise , Camundongos Knockout , Neurônios/química
7.
J Comp Neurol ; 527(7): 1210-1227, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30644550

RESUMO

Neurokinin B, encoded by the tachykinin3 gene, plays a crucial role in regulating reproduction in mammals via KNDy neurons and interaction with GnRH. Previous work in teleost fishes has focused on hypothalamic tac3 expression for its role in reproduction, but detailed studies on extra-hypothalamic tac3 expression are limited. Here, we identified two tac3 genes in the social African cichlid fish Astatotilapia burtoni, only one of which produces a functional protein containing the signature tachykinin motif. In situ hybridization for tac3a mRNA identified cell populations throughout the brain. Numerous tac3a cells lie in several thalamic and hypothalamic nuclei, including periventricular nucleus of posterior tuberculum, lateral tuberal nucleus (NLT), and nucleus of the lateral recess (NRL). Scattered tac3-expressing cells are also present in telencephalic parts, such as ventral (Vv) and supracomissural (Vs) part of ventral telencephalon. In contrast to other teleosts, tac3 expression was absent from the pituitary. Using double-fluorescent staining, we localized tac3a-expressing cells in relation to GnRH and kisspeptin cells. Although no GnRH-tac3a colabeled cells were observed, dense GnRH fibers surround and potentially synapse with tac3a cells in the preoptic area. Only minimal (<5%) colabeling of tac3a was observed in kiss2 cells. Despite tac3a expression in many nodes of the mesolimbic reward system, it was absent from tyrosine hydroxylase (TH)-expressing cells, but tac3a cells were located in areas with dense TH fibers. The presence of tac3a-expressing cells throughout the brain, including in socially relevant brain regions, suggest more diverse functions beyond regulation of reproductive physiology that may be conserved across vertebrates.


Assuntos
Encéfalo/metabolismo , Ciclídeos/metabolismo , Lectinas/biossíntese , Animais , Ciclídeos/genética , Neurônios Dopaminérgicos/fisiologia , Feminino , Peixes/classificação , Peixes/genética , Hormônio Liberador de Gonadotropina/análise , Hipotálamo/metabolismo , Hibridização In Situ , Kisspeptinas/análise , Lectinas/genética , Masculino , Especificidade de Órgãos , Filogenia , Reprodução/genética , Reprodução/fisiologia , Recompensa , Comportamento Social
8.
Clinics (Sao Paulo) ; 72(8): 510-514, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28954011

RESUMO

OBJECTIVES:: Polycystic ovary syndrome is a heterogeneous endocrine disorder that affects reproductive-age women. The mechanisms underlying the endocrine heterogeneity and neuroendocrinology of polycystic ovary syndrome are still unclear. In this study, we investigated the expression of the kisspeptin system and gonadotropin-releasing hormone pulse regulators in the hypothalamus as well as factors related to luteinizing hormone secretion in the pituitary of polycystic ovary syndrome rat models induced by testosterone or estradiol. METHODS:: A single injection of testosterone propionate (1.25 mg) (n=10) or estradiol benzoate (0.5 mg) (n=10) was administered to female rats at 2 days of age to induce experimental polycystic ovary syndrome. Controls were injected with a vehicle (n=10). Animals were euthanized at 90-94 days of age, and the hypothalamus and pituitary gland were used for gene expression analysis. RESULTS:: Rats exposed to testosterone exhibited increased transcriptional expression of the androgen receptor and estrogen receptor-ß and reduced expression of kisspeptin in the hypothalamus. However, rats exposed to estradiol did not show any significant changes in hormone levels relative to controls but exhibited hypothalamic downregulation of kisspeptin, tachykinin 3 and estrogen receptor-α genes and upregulation of the gene that encodes the kisspeptin receptor. CONCLUSIONS:: Testosterone- and estradiol-exposed rats with different endocrine phenotypes showed differential transcriptional expression of members of the kisspeptin system and sex steroid receptors in the hypothalamus. These differences might account for the different endocrine phenotypes found in testosterone- and estradiol-induced polycystic ovary syndrome rats.


Assuntos
Hormônio Liberador de Gonadotropina/análise , Hipotálamo/química , Kisspeptinas/análise , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Síndrome do Ovário Policístico/química , Animais , Modelos Animais de Doenças , Regulação para Baixo , Estradiol , Feminino , Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Fenótipo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/análise , Receptores de Estrogênio/análise , Testosterona , Regulação para Cima
9.
Clinics ; 72(8): 510-514, Aug. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-890718

RESUMO

OBJECTIVES: Polycystic ovary syndrome is a heterogeneous endocrine disorder that affects reproductive-age women. The mechanisms underlying the endocrine heterogeneity and neuroendocrinology of polycystic ovary syndrome are still unclear. In this study, we investigated the expression of the kisspeptin system and gonadotropin-releasing hormone pulse regulators in the hypothalamus as well as factors related to luteinizing hormone secretion in the pituitary of polycystic ovary syndrome rat models induced by testosterone or estradiol. METHODS: A single injection of testosterone propionate (1.25 mg) (n=10) or estradiol benzoate (0.5 mg) (n=10) was administered to female rats at 2 days of age to induce experimental polycystic ovary syndrome. Controls were injected with a vehicle (n=10). Animals were euthanized at 90-94 days of age, and the hypothalamus and pituitary gland were used for gene expression analysis. RESULTS: Rats exposed to testosterone exhibited increased transcriptional expression of the androgen receptor and estrogen receptor-β and reduced expression of kisspeptin in the hypothalamus. However, rats exposed to estradiol did not show any significant changes in hormone levels relative to controls but exhibited hypothalamic downregulation of kisspeptin, tachykinin 3 and estrogen receptor-α genes and upregulation of the gene that encodes the kisspeptin receptor. CONCLUSIONS: Testosterone- and estradiol-exposed rats with different endocrine phenotypes showed differential transcriptional expression of members of the kisspeptin system and sex steroid receptors in the hypothalamus. These differences might account for the different endocrine phenotypes found in testosterone- and estradiol-induced polycystic ovary syndrome rats.


Assuntos
Animais , Feminino , Hormônio Liberador de Gonadotropina/análise , Hipotálamo/química , Kisspeptinas/análise , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Síndrome do Ovário Policístico/química , Modelos Animais de Doenças , Regulação para Baixo , Estradiol , Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Fenótipo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/análise , Receptores de Estrogênio/análise , Testosterona , Regulação para Cima
10.
J Chin Med Assoc ; 79(10): 546-53, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27373140

RESUMO

BACKGROUND: The kisspeptin/kiss1r system, expressed in the hypothalamic arcuate nucleus, has been proclaimed as one of the most powerful factors of the reproduction axis, according to recent researches in the reproductive field. The aim of this study was to ascertain the expression of kisspeptin, its receptor (kiss1r), and gonadotropin-releasing hormone (GnRH), and to explore the role on the development and maintenance of the reproductive function of developing female rats. METHODS: Expressions of the kisspeptin/kiss1r system were examined by immunohistochemistry and Real time Quantitative PCR (qRT-PCR). Expressions of estradiol (E2), luteinizing hormone, and follicle-stimulating hormone were analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS: Expression of the kisspeptin/kiss1r system increased time dependently with aging, and their peak expression was demonstrated in the adult stage. GnRH showed a similar expression pattern to that of the kisspeptin/kiss1r system. ELISA results demonstrated that the E2, luteinizing hormone, and follicle-stimulating hormone secretion increased time dependently from infancy to prepuberty to puberty. However, E2 level decreased significantly in adult rats. Morphological changes of ovaries showed that primordial follicles, primary follicles, and growing follicle inhabited the dominant status in infancy, prepuberty, and puberty stages, respectively. CONCLUSION: GnRH neurons may play an intermediate role in the activation and maintenance of the reproductive function regulated by the kisspeptin/kiss1r system, which may also indirectly regulate the serum level of luteinizing hormone, follicle-stimulating hormone, and E2.


Assuntos
Hipotálamo/crescimento & desenvolvimento , Kisspeptinas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Reprodução/fisiologia , Animais , Estradiol/sangue , Estro/fisiologia , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/análise , Hormônio Liberador de Gonadotropina/fisiologia , Kisspeptinas/análise , Kisspeptinas/genética , Hormônio Luteinizante/sangue , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1
11.
Reprod Domest Anim ; 49(3): 433-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24716653

RESUMO

Normal reproductive function is dependent upon availability of glucose and insulin-induced hypoglycaemia is a metabolic stressor known to disrupt the ovine oestrous cycle. We have recently shown that IIH has the ability to delay the LH surge of intact ewes. In the present study, we examined brain tissue to determine: (i) which hypothalamic regions are activated with respect to IIH and (ii) the effect of IIH on kisspeptin cell activation and CRFR type 2 immunoreactivity, all of which may be involved in disruptive mechanisms. Follicular phases were synchronized with progesterone vaginal pessaries and at 28 h after progesterone withdrawal (PW), animals received saline (n = 6) or insulin (4 IU/kg; n = 5) and were subsequently killed at 31 h after PW (i.e., 3 h after insulin administration). Peripheral hormone concentrations were evaluated, and hypothalamic sections were immunostained for either kisspeptin and c-Fos (a marker of neuronal activation) or CRFR type 2. Within 3 h of treatment, cortisol concentrations had increased whereas plasma oestradiol concentrations decreased in peripheral plasma (p < 0.05 for both). In the arcuate nucleus (ARC), insulin-treated ewes had an increased expression of c-Fos. Furthermore, the percentage of kisspeptin cells co-expressing c-Fos increased in the ARC (from 11 to 51%; p < 0.05), but there was no change in the medial pre-optic area (mPOA; 14 vs 19%). CRFR type 2 expression in the lower part of the ARC and the median eminence was not altered by insulin treatment. Thus, disruption of the LH surge after IIH in the follicular phase is not associated with decreased kisspeptin cell activation or an increase in CRFR type 2 in the ARC but may involve other cell types located in the ARC nucleus which are activated in response to IIH.


Assuntos
Hipoglicemia/fisiopatologia , Hipotálamo/metabolismo , Kisspeptinas/genética , Proteínas Proto-Oncogênicas c-fos/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Ovinos/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/química , Núcleo Arqueado do Hipotálamo/fisiopatologia , Estradiol/sangue , Feminino , Fase Folicular/fisiologia , Regulação da Expressão Gênica , Hidrocortisona/sangue , Hipoglicemia/induzido quimicamente , Hipotálamo/química , Insulina/administração & dosagem , Kisspeptinas/análise , Hormônio Luteinizante/metabolismo , Progesterona/sangue , Proteínas Proto-Oncogênicas c-fos/análise , Reprodução/fisiologia
12.
Gen Comp Endocrinol ; 175(3): 432-42, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22166815

RESUMO

kisspeptins that are encoded by kiss1 gene are now considered the key regulator of reproduction from a number of studies in mammals. In most vertebrates, a paralogue of kiss1, called kiss2, is also present, and the functional significance of kisspeptins is not known precisely. In the present study, we have cloned kiss2 from a perciform teleost, the red seabream Pagrus major. The amino acid sequence deduced from the red seabream kiss2 contained a highly conserved 10-amino-acid residue, Kiss2(10) or kp-10. A kiss1-like transcript was also identified, but it appears to be non-functional due to the presence of a "premature" stop codon. Neurons that express kiss2 mRNA were distributed in the dorsal (NRLd) and ventral (NRLv) parts of nucleus recessi lateralis in the hypothalamus. In some fish a few kiss2-expressing neurons were detected in the preoptic area and nucleus ventralis tuberis. The number of kiss2-expressing neurons in the NRLd was larger during the first spawning season in both males and females compared with fish in the post-spawning periods. In males the number of kiss2 neurons in the NRLd of maturing fish was also larger than those in the post-spawning periods. In males the number of kiss2 neurons in the NRLv showed a similar pattern of changes to that of NRLd, while significant changes were not detected for females. The numbers of gonadotropin-releasing hormone 1 (GnRH1)-immunoreactive neurons in the preoptic area showed a similar pattern of change as those of kiss2 cells of the NRLd in both males and females (and also the NRLv in males). These results are in good agreement with the hypothesis that kiss2 neurons are involved in pubertal processes via regulatory influences on GnRH1 neurons in red seabream.


Assuntos
Encéfalo/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Dourada/fisiologia , Maturidade Sexual/fisiologia , Envelhecimento/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/citologia , Contagem de Células , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Hipotálamo/fisiologia , Kisspeptinas/análise , Kisspeptinas/genética , Masculino , Dados de Sequência Molecular , Neurônios/citologia , Área Pré-Óptica/fisiologia
13.
Neuroendocrinology ; 94(4): 323-32, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22116451

RESUMO

Kisspeptin has been thought to play pivotal roles in the control of both pulse and surge modes of gonadotropin-releasing hormone (GnRH) secretion. To clarify loci of kisspeptin action on GnRH neurons, the present study examined the morphology of the kisspeptin system and the associations between kisspeptin and GnRH systems in gonadally intact and castrated male goats. Kisspeptin-immunoreactive (ir) and Kiss1-positive neurons were found in the medial preoptic area of intact but not castrated goats. Kisspeptin-ir cell bodies and fibers in the arcuate nucleus (ARC) and median eminence (ME) were fewer in intact male goats compared with castrated animals. Apposition of kisspeptin-ir fibers on GnRH-ir cell bodies was very rare in both intact and castrated goats, whereas the intimate association of kisspeptin-ir fibers with GnRH-ir nerve terminals was observed in the ME of castrated animals. Neurokinin B immunoreactivity colocalized not only in kisspeptin-ir cell bodies in the ARC but also in kisspeptin-ir fibers in the ME, suggesting that a majority of kisspeptin-ir fibers projecting to the ME originates from the ARC. A dual immunoelectron microscopic examination revealed that nerve terminals containing kisspeptin-ir vesicles made direct contact with GnRH-ir nerve terminals at the ME of castrated goats. There was no evidence for the existence of the typical synaptic structure between kisspeptin- and GnRH-ir fibers. The present results suggest that the ARC kisspeptin neurons act on GnRH neurons at the ME to control (possibly the pulse mode of) GnRH secretion in males.


Assuntos
Hormônio Liberador de Gonadotropina/análise , Kisspeptinas/análise , Eminência Mediana/ultraestrutura , Neurônios/química , Animais , Núcleo Arqueado do Hipotálamo/química , Cabras , Hipotálamo/química , Imuno-Histoquímica , Masculino , Eminência Mediana/química , Eminência Mediana/citologia , Microscopia Imunoeletrônica , Neurocinina B/análise , Neurônios/ultraestrutura , Área Pré-Óptica/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA